1
|
Tang K, Ye T, He Y, Ba X, Xia D, Peng E, Chen Z, Ye Z, Yang X. Ferroptosis, necroptosis, and pyroptosis in calcium oxalate crystal-induced kidney injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167791. [PMID: 40086520 DOI: 10.1016/j.bbadis.2025.167791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Kidney stones represent a highly prevalent urological disorder worldwide, with high incidence and recurrence rates. Calcium oxalate (CaOx) crystal-induced kidney injury serves as the foundational mechanism for the formation and progression of CaOx stones. Regulated cell death (RCD) such as ferroptosis, necroptosis, and pyroptosis are essential in the pathophysiological process of kidney injury. Ferroptosis, a newly discovered RCD, is characterized by its reliance on iron-mediated lipid peroxidation. Necroptosis, a widely studied programmed necrosis, initiates with a necrotic phenotype that resembles apoptosis in appearance. Pyroptosis, a type of RCD that involves the gasdermin protein, is accompanied by inflammation and immune response. In recent years, increasing amounts of evidence has demonstrated that ferroptosis, necroptosis, and pyroptosis are significant pathophysiological processes involved in CaOx crystal-induced kidney injury. Herein, we summed up the roles of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury. Furthermore, we delved into the curative potential of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury.
Collapse
Affiliation(s)
- Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Elkhalil A, Whited A, Ghose P. SQST-1/p62-regulated SKN-1/Nrf mediates a phagocytic stress response via transcriptional activation of lyst-1/LYST. PLoS Genet 2025; 21:e1011696. [PMID: 40315422 PMCID: PMC12068719 DOI: 10.1371/journal.pgen.1011696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 05/12/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025] Open
Abstract
Cells may be intrinsically fated to die to sculpt tissues during development or to maintain homeostasis. Cells can also die in response to various stressors, injury or pathological conditions. Additionally, cells of the metazoan body are often highly specialized with distinct domains that differ both structurally and with respect to their neighbors. Specialized cells can also die, as in normal brain development or pathological states and their different regions may be eliminated via different programs. Clearance of different types of cell debris must be performed quickly and efficiently to prevent autoimmunity and secondary necrosis of neighboring cells. Moreover, all cells, including those programmed to die, may be subject to various stressors. Some largely unexplored questions include whether predestined cell elimination during development could be altered by stress, if adaptive stress responses exist and if polarized cells may need compartment-specific stress-adaptive programs. We leveraged Compartmentalized Cell Elimination (CCE) in the nematode C. elegans to explore these questions. CCE is a developmental cell death program whereby three segments of two embryonic polarized cell types are eliminated differently. We have previously employed this in vivo genetic system to uncover a cell compartment-specific, cell non-autonomous clearance function of the fusogen EFF-1 in phagosome closure during corpse internalization. Here, we introduce an adaptive response that serves to aid developmental phagocytosis as a part of CCE during stress. We employ a combination of forward and reverse genetics, CRISPR/Cas9 gene editing, stress response assays and advanced fluorescence microscopy. Specifically, we report that, under heat stress, the selective autophagy receptor SQST-1/p62 promotes the nuclear translocation of the oxidative stress-related transcription factor SKN-1/Nrf via negative regulation of WDR-23. This in turn allows SKN-1/Nrf to transcribe lyst-1/LYST (lysosomal trafficking associated gene) which subsequently promotes the phagocytic resolution of the developmentally-killed internalized cell even under stress conditions.
Collapse
Affiliation(s)
- Aladin Elkhalil
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Alec Whited
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Piya Ghose
- The University of Texas at Arlington, Arlington, Texas, United States of America
| |
Collapse
|
3
|
Li W, Deng K, Zhang M, Xu Y, Zhang J, Liang Q, Yang Z, Jin L, Hu C, Zhao YT. Network Pharmacology Combined with Experimental Validation to Investigate the Effects and Mechanisms of Aucubin on Aging-Related Muscle Atrophy. Int J Mol Sci 2025; 26:2626. [PMID: 40141269 PMCID: PMC11941843 DOI: 10.3390/ijms26062626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Aucubin (AU) is one of the main components of the traditional Chinese medicine Eucommia ulmoides Oliv (EU). This study investigated the effects of AU on aging-related skeletal muscle atrophy in vitro and in vivo. The results of network pharmacology revealed the potential therapeutic effects of AU on muscle atrophy. In vitro, AU effectively attenuated D-gal-induced cellular damage, reduced the number of senescence-associated β-galactosidase (SA-β-Gal)-positive cells, down-regulated the expression levels of muscle atrophy-related proteins Atrogin-1 and MuRF1, and improved myotube differentiation, thereby mitigating myotube atrophy. Notably, AU was found to attenuate oxidative stress and apoptosis in skeletal muscle cells by reducing ROS production, regulating Cleaved caspase3 and BAX/Bcl-2 expression in apoptotic pathways, and enhancing Sirt1 and PGC-1α signaling pathways. In vivo studies demonstrated that AU treatment extended the average lifespan of Caenorhabditis elegans (C. elegans), increased locomotor activity, improved body wall muscle mitochondrial content, and alleviated oxidative damage in C. elegans. These findings suggested that AU can ameliorate aging-related muscle atrophy and show significant potential in preventing and treating muscle atrophy.
Collapse
Affiliation(s)
- Wenan Li
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| | - Kaishu Deng
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| | - Mengyue Zhang
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| | - Yan Xu
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| | - Jingxi Zhang
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| | - Qingsheng Liang
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| | - Zhiyou Yang
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| | - Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China;
| | - Chuanyin Hu
- Department of Biology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yun-Tao Zhao
- Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Modern Biochemistry Experimental Center, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Guangdong Ocean University, Zhanjiang 524088, China (K.D.); (M.Z.); (Y.X.); (J.Z.); (Q.L.); (Z.Y.)
| |
Collapse
|
4
|
Liu Y, Cao Y, Li H, Liu H, Chen T, Lin Q, Gong C, Yu F, Cai H, Jin L, Peng R. Mitochondrial homeostatic imbalance-mediated developmental toxicity to H 2S in embryonic zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125588. [PMID: 39725203 DOI: 10.1016/j.envpol.2024.125588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/01/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024]
Abstract
Hydrogen sulfide (H2S) is a pervasive environmental and industrial pollutant that poses a substantial threat to human health. Even short-term exposure to H2S can result in severe respiratory and neurological damage. However, the underlying mechanisms of its biotoxicity remain unclear. Our study demonstrated that continuous exposure to 30 μM (1.02 ppm), whin environmentally H2S concentration range, results in notable developmental toxicity, including high mortality rates, morphological deformities, and behavioral abnormalities, in zebrafish larvae. Through transcriptomic analysis, examination of mitochondrial structure and function, and tissue and cellular staining, we found that H2S exposure disrupted mitochondrial dynamics, autophagy, and biogenesis, leading to an imbalance in mitochondrial homeostasis. This disruption induced oxidative stress and extensive apoptosis. Nitric oxide (NO) is a multifunctional signaling molecule known to target and regulate mitochondrial regeneration. In our study, we discovered that sodium nitroprusside (SNP), an NO donor, can activate the NO-sGC-cGMP signaling pathway. This activation improves the homeostatic regulation of mitochondrial dynamics, autophagy, and biogenesis, thereby enhancing mitochondrial function and effectively mitigating H2S-induced biotoxicity. Our research not only elucidates the biotoxicity mechanisms of H2S exposure but also provides valuable insights into potential therapeutic strategies that alleviate or eliminate its toxic effects.
Collapse
Affiliation(s)
- Yinai Liu
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yu Cao
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Huiqi Li
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Huanpeng Liu
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Ting Chen
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Qizhuan Lin
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Changyong Gong
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Fan Yu
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Helei Cai
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Libo Jin
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| |
Collapse
|
5
|
Horowitz LB, Shaham S. Apoptotic and Nonapoptotic Cell Death in Caenorhabditis elegans Development. Annu Rev Genet 2024; 58:113-134. [PMID: 38955209 DOI: 10.1146/annurev-genet-111523-102051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Programmed cell death (PCD) is an essential component of animal development, and aberrant cell death underlies many disorders. Understanding mechanisms that govern PCD during development can provide insight into cell death programs that are disrupted in disease. Key steps mediating apoptosis, a highly conserved cell death program employing caspase proteases, were first uncovered in the nematode Caenorhabditis elegans, a powerful model system for PCD research. Recent studies in C. elegans also unearthed conserved nonapoptotic caspase-independent cell death programs that function during development. Here, we discuss recent advances in understanding cell death during C. elegans development. We review insights expanding the molecular palette behind the execution of apoptotic and nonapoptotic cell death, as well as new discoveries revealing the mechanistic underpinnings of dying cell engulfment and clearance. A number of open questions are also discussed that will continue to propel the field over the coming years.
Collapse
Affiliation(s)
- Lauren Bayer Horowitz
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA; ,
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY, USA; ,
| |
Collapse
|
6
|
Damilou A, Cai L, Argunşah AÖ, Han S, Kanatouris G, Karatsoli M, Hanley O, Gesuita L, Kollmorgen S, Helmchen F, Karayannis T. Developmental Cajal-Retzius cell death contributes to the maturation of layer 1 cortical inhibition and somatosensory processing. Nat Commun 2024; 15:6501. [PMID: 39090081 PMCID: PMC11294614 DOI: 10.1038/s41467-024-50658-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 07/11/2024] [Indexed: 08/04/2024] Open
Abstract
The role of developmental cell death in the formation of brain circuits is not well understood. Cajal-Retzius cells constitute a major transient neuronal population in the mammalian neocortex, which largely disappears at the time of postnatal somatosensory maturation. In this study, we used mouse genetics, anatomical, functional, and behavioral approaches to explore the impact of the early postnatal death of Cajal-Retzius cells in the maturation of the cortical circuit. We find that before their death, Cajal-Retzius cells mainly receive inputs from layer 1 neurons, which can only develop their mature connectivity onto layer 2/3 pyramidal cells after Cajal-Retzius cells disappear. This developmental connectivity progression from layer 1 GABAergic to layer 2/3 pyramidal cells regulates sensory-driven inhibition within, and more so, across cortical columns. Here we show that Cajal-Retzius cell death prevention leads to layer 2/3 hyper-excitability, delayed learning and reduced performance in a multi-whisker-dependent texture discrimination task.
Collapse
Affiliation(s)
- Angeliki Damilou
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich, 8057, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Linbi Cai
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Ali Özgür Argunşah
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Shuting Han
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - George Kanatouris
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Maria Karatsoli
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich, 8057, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Olivia Hanley
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Lorenzo Gesuita
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Sepp Kollmorgen
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich, 8057, Switzerland
| | - Fritjof Helmchen
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich, 8057, Switzerland
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Theofanis Karayannis
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich, 8057, Switzerland.
- Neuroscience Center Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
7
|
Berry CW, Fuller MT. Functional septate junctions between cyst cells are required for survival of transit amplifying male germ cells expressing Bag of marbles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587826. [PMID: 38617328 PMCID: PMC11014526 DOI: 10.1101/2024.04.02.587826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
In adult stem cell lineages, the cellular microenvironment plays essential roles to ensure the proper balance of self-renewal, differentiation and regulated elimination of differentiating cells. Although regulated death of progenitor cells undergoing proliferation or early differentiation is a feature of many tissues, mechanisms that initiate this pruning remain unexplored, particularly in the male germline, where up to 30% of the germline is eliminated before the meiotic divisions. We conducted a targeted screen to identify functional regulators required in somatic support cells for survival or differentiation at early steps in the male germ line stem cell lineage. Cell type-specific knockdown in cyst cells uncovered novel roles of genes in germline stem cell differentiation, including a previously unappreciated role of the Septate Junction (SJ) in preventing cell death of differentiating germline progenitors. Loss of the SJ in the somatic cyst cells resulted in elimination of transit-amplifying spermatogonia by the 8-cell stage. Germ cell death was spared in males mutant for the differentiation factor bam indicating that intact barriers surrounding transit amplifying progenitors are required to ensure germline survival once differentiation has initiated.
Collapse
Affiliation(s)
- Cameron W. Berry
- Department of Developmental Biology, Stanford University School of Medicine, USA
| | - Margaret T. Fuller
- Department of Developmental Biology, Stanford University School of Medicine, USA
- Department of Genetics, Stanford University School of Medicine, USA
| |
Collapse
|
8
|
Yarychkivska O, Sharmin R, Elkhalil A, Ghose P. Apoptosis and beyond: A new era for programmed cell death in Caenorhabditis elegans. Semin Cell Dev Biol 2024; 154:14-22. [PMID: 36792437 DOI: 10.1016/j.semcdb.2023.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
Programmed cell death (PCD) is crucial for normal development and homeostasis. Our first insights into the genetic regulation of apoptotic cell death came from in vivo studies in the powerful genetic model system of C. elegans. More recently, novel developmental cell death programs occurring both embryonically and post-embryonically, and sex-specifically, have been elucidated. Recent studies in the apoptotic setting have also shed new light on the intricacies of phagocytosis in particular. This review provides a brief historical perspective of the origins of PCD studies in C. elegans, followed by a more detailed description of non-canonical apoptotic and non-apoptotic death programs. We conclude by posing open questions and commenting on our outlook on the future of PCD studies in C. elegans, highlighting the importance of advanced imaging tools and the continued leveraging of C. elegans genetics both with classical and modern cutting-edge approaches.
Collapse
Affiliation(s)
| | | | | | - Piya Ghose
- The University of Texas at Arlington, USA.
| |
Collapse
|
9
|
Wang H, Wang X, Huang L, Wang C, Yu F, Ye L. Overburdened ferroptotic stress impairs tooth morphogenesis. eLife 2023; 12:RP88745. [PMID: 37991825 PMCID: PMC10665014 DOI: 10.7554/elife.88745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
The role of regulated cell death in organ development, particularly the impact of non-apoptotic cell death, remains largely uncharted. Ferroptosis, a non-apoptotic cell death pathway known for its iron dependence and lethal lipid peroxidation, is currently being rigorously investigated for its pathological functions. The balance between ferroptotic stress (iron and iron-dependent lipid peroxidation) and ferroptosis supervising pathways (anti-lipid peroxidation systems) serves as the key mechanism regulating the activation of ferroptosis. Compared with other forms of regulated necrotic cell death, ferroptosis is critically related to the metabolism of lipid and iron which are also important in organ development. In our study, we examined the role of ferroptosis in organogenesis using an ex vivo tooth germ culture model, investigating the presence and impact of ferroptotic stress on tooth germ development. Our findings revealed that ferroptotic stress increased during tooth development, while the expression of glutathione peroxidase 4 (Gpx4), a crucial anti-lipid peroxidation enzyme, also escalated in dental epithelium/mesenchyme cells. The inhibition of ferroptosis was found to partially rescue erastin-impaired tooth morphogenesis. Our results suggest that while ferroptotic stress is present during tooth organogenesis, its effects are efficaciously controlled by the subsequent upregulation of Gpx4. Notably, an overabundance of ferroptotic stress, as induced by erastin, suppresses tooth morphogenesis.
Collapse
Affiliation(s)
- Haisheng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengduChina
| | - Xiaofeng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengduChina
- Department of Endodontics, West China School of Stomatology, Sichuan UniversityChengduChina
| | - Liuyan Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengduChina
- Department of Endodontics, West China School of Stomatology, Sichuan UniversityChengduChina
| | - Chenglin Wang
- Department of Endodontics, West China School of Stomatology, Sichuan UniversityChengduChina
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengduChina
- Department of Endodontics, West China School of Stomatology, Sichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengduChina
- Department of Endodontics, West China School of Stomatology, Sichuan UniversityChengduChina
| |
Collapse
|
10
|
Packard M, Gilbert MC, Tetrault E, Albertson RC. Zebrafish crocc2 mutants exhibit divergent craniofacial shape, misregulated variability, and aberrant cartilage morphogenesis. Dev Dyn 2023; 252:1026-1045. [PMID: 37032317 PMCID: PMC10524572 DOI: 10.1002/dvdy.591] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/11/2023] Open
Abstract
BACKGROUND Phenotypic variation is of paramount importance in development, evolution, and human health; however, the molecular mechanisms that influence organ shape and shape variability are not well understood. During craniofacial development, the behavior of skeletal precursors is regulated by both biochemical and environmental inputs, and the primary cilia play critical roles in transducing both types of signals. Here, we examine a gene that encodes a key constituent of the ciliary rootlets, crocc2, and its role in cartilage morphogenesis in larval zebrafish. RESULTS Geometric morphometric analysis of crocc2 mutants revealed altered craniofacial shapes and expanded variation. At the cellular level, we observed altered chondrocyte shapes and planar cell polarity across multiple stages in crocc2 mutants. Notably, cellular defects were specific to areas that experience direct mechanical input. Cartilage cell number, apoptosis, and bone patterning were not affected in crocc2 mutants. CONCLUSIONS Whereas "regulatory" genes are widely implicated in patterning the craniofacial skeleton, genes that encode "structural" aspects of the cell are increasingly implicated in shaping the face. Our results add crocc2 to this list, and demonstrate that it affects craniofacial geometry and canalizes phenotypic variation. We propose that it does so via mechanosensing, possibly through the ciliary rootlet. If true, this would implicate a new organelle in skeletal development and evolution.
Collapse
Affiliation(s)
- Mary Packard
- Department of Biology, University of Massachusetts, Amherst, MA 01003, U.S.A
| | - Michelle C. Gilbert
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, U.S.A
- Current address, Department of Biology, Penn State University, University Park, PA 16802, U.S.A
| | - Emily Tetrault
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, U.S.A
| | - R. Craig Albertson
- Department of Biology, University of Massachusetts, Amherst, MA 01003, U.S.A
| |
Collapse
|
11
|
Fu Y, He Y, Wu D, Sui B, Jin Y, Hu X, Shi S. Apoptotic vesicles: emerging concepts and research progress in physiology and therapy. LIFE MEDICINE 2023; 2:lnad013. [PMID: 39872110 PMCID: PMC11749838 DOI: 10.1093/lifemedi/lnad013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/25/2022] [Indexed: 01/29/2025]
Abstract
Apoptosis represents the dominant form of programmed cell death and plays critical roles in maintaining tissue and organ homeostasis. A notable population of extracellular vesicles (EVs) is generated during apoptosis, known as apoptotic vesicles (apoVs). These apoVs are increasingly the subject of studies concerning their identity and mechanisms of production, which have been revealed unique biological and functional characteristics that are emerging as crucial regulators for diverse processes. Furthermore, apoVs have been gradually noticed for their essential role in regulating the physiology of various organ systems in vivo, and growing evidence suggests that apoV dysregulation contributes to age- and pathology-associated tissue alterations. Importantly, apoVs can be therapeutically harnessed to unleash their potential in treating several diseases such as immune disorders, osteoporosis, cutaneous wound and acute liver failure; these vesicles, mainly derived from cultured mesenchymal stem cells, hold great translational promise. Here we review the current landscape of scientific knowledge about apoVs, with emphasis on mechanistic insights into how apoVs contribute to organismal health and disease, which also provide novel cell-free strategies for EV-based regenerative therapeutics.
Collapse
Affiliation(s)
- Yu Fu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Fujian Key Laboratory of Developmental and Neural Biology and Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Di Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Bingdong Sui
- Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi’an 710032, China
| | - Yan Jin
- Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi’an 710032, China
| | - Xuefeng Hu
- Fujian Key Laboratory of Developmental and Neural Biology and Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
12
|
Petanjek Z, Banovac I, Sedmak D, Hladnik A. Dendritic Spines: Synaptogenesis and Synaptic Pruning for the Developmental Organization of Brain Circuits. ADVANCES IN NEUROBIOLOGY 2023; 34:143-221. [PMID: 37962796 DOI: 10.1007/978-3-031-36159-3_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synaptic overproduction and elimination is a regular developmental event in the mammalian brain. In the cerebral cortex, synaptic overproduction is almost exclusively correlated with glutamatergic synapses located on dendritic spines. Therefore, analysis of changes in spine density on different parts of the dendritic tree in identified classes of principal neurons could provide insight into developmental reorganization of specific microcircuits.The activity-dependent stabilization and selective elimination of the initially overproduced synapses is a major mechanism for generating diversity of neural connections beyond their genetic determination. The largest number of overproduced synapses was found in the monkey and human cerebral cortex. The highest (exceeding adult values by two- to threefold) and most protracted overproduction (up to third decade of life) was described for associative layer IIIC pyramidal neurons in the human dorsolateral prefrontal cortex.Therefore, the highest proportion and extraordinarily extended phase of synaptic spine overproduction is a hallmark of neural circuitry in human higher-order associative areas. This indicates that microcircuits processing the most complex human cognitive functions have the highest level of developmental plasticity. This finding is the backbone for understanding the effect of environmental impact on the development of the most complex, human-specific cognitive and emotional capacities, and on the late onset of human-specific neuropsychiatric disorders, such as autism and schizophrenia.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
13
|
Reddy CN, Sankararamakrishnan R. Molecular dynamics studies of CED-4/CED-9/EGL-1 ternary complex reveal CED-4 release mechanism in the linear apoptotic pathway of Caenorhabditis elegans. Proteins 2022; 91:679-693. [PMID: 36541866 DOI: 10.1002/prot.26457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/14/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Many steps in programmed cell death are evolutionarily conserved across different species. The Caenorhabditis elegans proteins CED-9, CED-4 and EGL-1 involved in apoptosis are respectively homologous to anti-apoptotic Bcl-2 proteins, Apaf-1 and the "BH3-only" pro-apototic proteins in mammals. In the linear apoptotic pathway of C. elegans, EGL-1 binding to CED-9 leads to the release of CED-4 from CED-9/CED-4 complex. The molecular events leading to this process are not clearly elucidated. While the structures of CED-9 apo, CED-9/EGL-1 and CED-9/CED-4 complexes are known, the CED-9/CED-4/EGL-1 ternary complex structure is not yet determined. In this work, we modeled this ternary complex and performed molecular dynamics simulations of six different systems involving CED-9. CED-9 displays differential dynamics depending upon whether it is bound to CED-4 and/or EGL-1. CED-4 exists as an asymmetric dimer (CED4a and CED4b) in CED-9/CED-4 complex. CED-4a exhibits higher conformational flexibility when simulated without CED-4b. Principal Component Analysis revealed that the direction of CED-4a's winged-helix domain motion differs in the ternary complex. Upon EGL-1 binding, majority of non-covalent interactions involving CARD domain in the CED-4a-CED-9 interface have weakened and only half of the contacts found in the crystal structure between α/β domain of CED4a and CED-9 are found to be stable. Additional stable contacts in the ternary complex and differential dynamics indicate that winged-helix domain may play a key role in CED-4a's dissociation from CED-9. This study has provided a molecular level understanding of potential intermediate states that are likely to occur when CED-4a is released from CED-9.
Collapse
Affiliation(s)
- C Narendra Reddy
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, India
| | - Ramasubbu Sankararamakrishnan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, India.,Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
14
|
Life-Saver or Undertaker: The Relationship between Primary Cilia and Cell Death in Vertebrate Embryonic Development. J Dev Biol 2022; 10:jdb10040052. [PMID: 36547474 PMCID: PMC9783631 DOI: 10.3390/jdb10040052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The development of multicellular organisms requires a tightly coordinated network of cellular processes and intercellular signalling. For more than 20 years, it has been known that primary cilia are deeply involved in the mediation of intercellular signalling and that ciliary dysfunction results in severe developmental defects. Cilia-mediated signalling regulates cellular processes such as proliferation, differentiation, migration, etc. Another cellular process ensuring proper embryonic development is cell death. While the effect of cilia-mediated signalling on many cellular processes has been extensively studied, the relationship between primary cilia and cell death remains largely unknown. This article provides a short review on the current knowledge about this relationship.
Collapse
|
15
|
Song C, Broadie K. Dysregulation of BMP, Wnt, and Insulin Signaling in Fragile X Syndrome. Front Cell Dev Biol 2022; 10:934662. [PMID: 35880195 PMCID: PMC9307498 DOI: 10.3389/fcell.2022.934662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/06/2022] [Indexed: 01/21/2023] Open
Abstract
Drosophila models of neurological disease contribute tremendously to research progress due to the high conservation of human disease genes, the powerful and sophisticated genetic toolkit, and the rapid generation time. Fragile X syndrome (FXS) is the most prevalent heritable cause of intellectual disability and autism spectrum disorders, and the Drosophila FXS disease model has been critical for the genetic screening discovery of new intercellular secretion mechanisms. Here, we focus on the roles of three major signaling pathways: BMP, Wnt, and insulin-like peptides. We present Drosophila FXS model defects compared to mouse models in stem cells/embryos, the glutamatergic neuromuscular junction (NMJ) synapse model, and the developing adult brain. All three of these secreted signaling pathways are strikingly altered in FXS disease models, giving new mechanistic insights into impaired cellular outcomes and neurological phenotypes. Drosophila provides a powerful genetic screening platform to expand understanding of these secretory mechanisms and to test cellular roles in both peripheral and central nervous systems. The studies demonstrate the importance of exploring broad genetic interactions and unexpected regulatory mechanisms. We discuss a number of research avenues to pursue BMP, Wnt, and insulin signaling in future FXS investigations and the development of potential therapeutics.
Collapse
Affiliation(s)
- Chunzhu Song
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
| | - Kendal Broadie
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
- Department of Cell and Developmental Biology, School of Medicine, Vanderbilt University, Nashville, TN, United States
- Kennedy Center for Research on Human Development, Nashville, TN, United States
- Vanderbilt Brain Institute, School of Medicine, Vanderbilt University and Medical Center, Nashville, TN, United States
| |
Collapse
|
16
|
Zuo Y, Shen H, Sun F, Li P, Sun J, Kwok RTK, Lam JWY, Tang BZ. Aggregation-Induced Emission Luminogens for Cell Death Research. ACS BIO & MED CHEM AU 2022; 2:236-257. [PMID: 37101570 PMCID: PMC10114857 DOI: 10.1021/acsbiomedchemau.1c00066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Cell death is closely related to various diseases, and monitoring and controlling cell death is a promising strategy to develop efficient therapy. Aggregation-induced emission luminogens (AIEgens) are ideal candidates for developing novel theranostic agents because of their intriguing properties in the aggregate state. The rational application of AIE materials in cell death-related research is still in its infancy but has shown great clinical potential. This review discussed the research frontier and our understanding of AIE materials in various subroutines of cell death, including apoptosis, necrosis, immunogenic cell death, pyroptosis, autophagy, lysosome-dependent cell death, and ferroptosis. We hope that the new insights can be offered to this growing field and attract more researchers to provide valuable contributions.
Collapse
Affiliation(s)
- Yunfei Zuo
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Hanchen Shen
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Feiyi Sun
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Pei Li
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
- Department
of Gastrointestinal Surgery, The Second Clinical Medical College, Shenzhen People’s Hospital, Jinan University, Shenzhen, 518020, China
| | - Jianwei Sun
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Ryan T. K. Kwok
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Jacky W. Y. Lam
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Ben Zhong Tang
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
- Shenzhen
Institute of Aggregate Science and Technology, School of Science and
Engineering, The Chinese University of Hong
Kong, Shenzhen, 2001
Longxiang Boulevard, Longgang District, Shenzhen
City, Guangdong 518172, China
| |
Collapse
|
17
|
The Leaf Extract of Mitrephora chulabhorniana Suppresses Migration and Invasion and Induces Human Cervical Cancer Cell Apoptosis through Caspase-Dependent Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2028082. [PMID: 35655474 PMCID: PMC9152413 DOI: 10.1155/2022/2028082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 12/20/2022]
Abstract
Cervical cancer is rated to be the leading cause of cancer-related death in women worldwide. Since screening test and conventional treatments are less accessible for people in developing countries, an alternative use of medicinal plants exhibiting strong anticancer activities may be an affordable means to treat cervical cancer. Mitrephora chulabhorniana (MC) is the newly identified species; however, its biological functions including anticancer activities have been largely unexplored. Hence, in this study, we were interested in investigating anticancer effects of this plant on the human cervical cell line (HeLa). MC extract was profiled for phytochemicals by TLC. This plant was tested to contain alkaloids, flavonoids, and terpenes. HeLa cells were treated with MC extract to investigate the anticancer activities. Cytotoxicity and viability of cells treated with MC were determined by MTT assay and Trypan blue exclusion assay. Cell migration was tested by wound healing assay, and cell invasion was determined by Transwell assay. The level of caspase 7, caspase 9, and PARP was determined by western blot analysis. We found that the leaf extract of MC strongly reduced cancer cell survival rate. This finding was consistent with the discovery that the extract dramatically induced apoptosis of cervical cancer cells through the activation of caspase 7 and caspase 9 which consequently degraded PARP protein. Furthermore, MC extract at lower concentrations which were not cytotoxic to the cancer cells showed potent inhibitory activities against HeLa cervical cancer cell migration and invasion. Mitrephora chulabhorniana possesses its pharmacological properties in inhibiting cervical cancer cell migration/invasion and inducing apoptotic signaling. This accumulated information suggests that Mitrephora chulabhorniana may be a beneficial source of potential agents for cervical cancer treatment.
Collapse
|
18
|
Xiong L, Bin Zhou, Young JL, Wintergerst K, Cai L. Exposure to low-dose cadmium induces testicular ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 234:113373. [PMID: 35272187 PMCID: PMC10858319 DOI: 10.1016/j.ecoenv.2022.113373] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
As an environmental pollutant, cadmium (Cd) has been widely reported to induce male infertility due to its gonadotoxicity. However, the specific mechanism of Cd-induced testicular damage remains unclear. We investigated whether Cd causes testicular injury through ferroptosis. Male C57BL/6 J mice were exposed to 0, 0.5, or 5 ppm Cd via drinking water, starting in utero, and continuing through 24 weeks post-weaning. The results showed that Cd accumulated in the testes in a dose-dependent manner. Cd exposure at a concentration of 5 ppm, but not 0.5 ppm, caused a mass loss and detachment of germ cells, as well as a decreased meiotic index and testis weight. Exposure to 5 ppm Cd caused iron accumulation, increased levels of malondialdehyde (MDA) and nitro tyrosine (3-NT), and decreased expression of Nrf2, HO-1 and SOD2. We also found that exposure to 5 ppm Cd significantly decreased the expression of SLC7A11, a marker of ferroptosis in mice, along with the expression of SLC40A1 mRNA and ferritin heavy chain (FTH) protein, whereas there was no obvious change in the mRNA expression of Tfrc, ZIP8, ZIP14, and NCOA4. These findings indicate that 5 ppm Cd exposure increased testicular ferroptosis, which may be attributed to the reduction of stored iron export.
Collapse
Affiliation(s)
- Lijuan Xiong
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Emergency, The Affiliated Children's Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Bin Zhou
- Department of Endocrinology, Metabolism, and Genetics, The Affiliated Children's Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jamie L Young
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kupper Wintergerst
- Wendy Novak Diabetes Center, Norton Children's Hospital, Louisville, KY 40202, USA; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; Wendy Novak Diabetes Center, Norton Children's Hospital, Louisville, KY 40202, USA; Radiation Oncology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
19
|
Juanez K, Ghose P. Repurposing the Killing Machine: Non-canonical Roles of the Cell Death Apparatus in Caenorhabditis elegans Neurons. Front Cell Dev Biol 2022; 10:825124. [PMID: 35237604 PMCID: PMC8882910 DOI: 10.3389/fcell.2022.825124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
Here we highlight the increasingly divergent functions of the Caenorhabditis elegans cell elimination genes in the nervous system, beyond their well-documented roles in cell dismantling and removal. We describe relevant background on the C. elegans nervous system together with the apoptotic cell death and engulfment pathways, highlighting pioneering work in C. elegans. We discuss in detail the unexpected, atypical roles of cell elimination genes in various aspects of neuronal development, response and function. This includes the regulation of cell division, pruning, axon regeneration, and behavioral outputs. We share our outlook on expanding our thinking as to what cell elimination genes can do and noting their versatility. We speculate on the existence of novel genes downstream and upstream of the canonical cell death pathways relevant to neuronal biology. We also propose future directions emphasizing the exploration of the roles of cell death genes in pruning and guidance during embryonic development.
Collapse
|
20
|
Liu J, Zhu S, Zeng L, Li J, Klionsky DJ, Kroemer G, Jiang J, Tang D, Kang R. DCN released from ferroptotic cells ignites AGER-dependent immune responses. Autophagy 2021; 18:2036-2049. [PMID: 34964698 PMCID: PMC9397459 DOI: 10.1080/15548627.2021.2008692] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis is a form of inflammatory cell death for which key mediators remain obscure. Here, we report that the proteoglycan decorin (DCN) is released by cells that are dying from ferroptosis and then acts as an alarm signal to trigger innate and adaptive immune responses. The early release of DCN during ferroptosis is an active process that involves secretory macroautophagy/autophagy and lysosomal exocytosis. Once released, extracellular DCN binds to the receptor advanced glycosylation end-product-specific receptor (AGER) on macrophages to trigger the production of pro-inflammatory cytokines in an NFKB/NF-κB-dependent manner. Pharmacological and genetic inhibition of the DCN-AGER axis protects against ferroptotic death-related acute pancreatitis and limits the capacity of ferroptotic cancer cells to induce a tumor-protective immune response. Thus, DCN is an essential mediator of the inflammatory and immune consequences of ferroptosis.
Collapse
Affiliation(s)
- Jiao Liu
- Center for DAMP Biology, Third Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Shan Zhu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Chongqing, China
| | - Jingbo Li
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Chongqing, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
21
|
Compagnucci C, Martinus K, Griffin J, Depew MJ. Programmed Cell Death Not as Sledgehammer but as Chisel: Apoptosis in Normal and Abnormal Craniofacial Patterning and Development. Front Cell Dev Biol 2021; 9:717404. [PMID: 34692678 PMCID: PMC8531503 DOI: 10.3389/fcell.2021.717404] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/28/2021] [Indexed: 12/22/2022] Open
Abstract
Coordination of craniofacial development involves an complex, intricate, genetically controlled and tightly regulated spatiotemporal series of reciprocal inductive and responsive interactions among the embryonic cephalic epithelia (both endodermal and ectodermal) and the cephalic mesenchyme — particularly the cranial neural crest (CNC). The coordinated regulation of these interactions is critical both ontogenetically and evolutionarily, and the clinical importance and mechanistic sensitivity to perturbation of this developmental system is reflected by the fact that one-third of all human congenital malformations affect the head and face. Here, we focus on one element of this elaborate process, apoptotic cell death, and its role in normal and abnormal craniofacial development. We highlight four themes in the temporospatial elaboration of craniofacial apoptosis during development, namely its occurrence at (1) positions of epithelial-epithelial apposition, (2) within intra-epithelial morphogenesis, (3) during epithelial compartmentalization, and (4) with CNC metameric organization. Using the genetic perturbation of Satb2, Pbx1/2, Fgf8, and Foxg1 as exemplars, we examine the role of apoptosis in the elaboration of jaw modules, the evolution and elaboration of the lambdoidal junction, the developmental integration at the mandibular arch hinge, and the control of upper jaw identity, patterning and development. Lastly, we posit that apoptosis uniquely acts during craniofacial development to control patterning cues emanating from core organizing centres.
Collapse
Affiliation(s)
- Claudia Compagnucci
- Institute for Cell and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, CCO, Berlin, Germany.,Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy.,Department of Craniofacial Development, King's College London, London, United Kingdom
| | - Kira Martinus
- Institute for Cell and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, CCO, Berlin, Germany
| | - John Griffin
- Department of Craniofacial Development, King's College London, London, United Kingdom.,School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Michael J Depew
- Institute for Cell and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, CCO, Berlin, Germany.,Department of Craniofacial Development, King's College London, London, United Kingdom
| |
Collapse
|
22
|
Early cellular development induced by ecdysteroid in sex-specific wing degeneration of the wingless female winter moth. Cell Tissue Res 2021; 387:29-38. [PMID: 34661757 DOI: 10.1007/s00441-021-03540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/08/2021] [Indexed: 10/20/2022]
Abstract
The winter moth, Nyssiodes lefuarius, exhibits striking sexual dimorphism in wing form; males have functional wings of normal size, whereas females lack wings. We previously found that the steroid hormone 20-hydroxyecdysone (20E) triggered massive programmed cell death (PCD) only in the female pupal wing epithelium; however, when and how early sexual trait development of the pupal wings is initiated during pupal-adult metamorphosis remains obscure. To clarify the detailed morphological changes and mechanisms underlying early sexual trait development and cell death, we examined the effects of 20E on early ultrastructural and histological changes in the pupal wing epithelium of both sexes. Before the onset of adult differentiation, no morphological differences were observed in the epithelial cells of both sexes at an ultrastructural level. When 5.4 µg of 20E was injected into pupae of both sexes at 15 days after the onset of pupation, retraction of the wing epithelium from the pupal cuticle was initiated at day 2 after 20E injection in both sexes. Although overt degeneration of wing tissue was not still obvious, apoptotic body-like structures and auto-phagosomes were visible at day 3 after 20E injection in females, whereas development of scale precursor cells started on day 4 after injection in males. Our results suggest that (1) the injection of 20E induced sexually dimorphic changes in the pattern of organelle distribution in wing epithelial cells, and (2) abnormally shaped mitochondria in the cytoplasm of the female wing epithelium might be involved in the PCD that occurs during wing tissue degeneration.
Collapse
|
23
|
Orchestration of Force Generation and Nuclear Collapse in Apoptotic Cells. Int J Mol Sci 2021; 22:ijms221910257. [PMID: 34638598 PMCID: PMC8508646 DOI: 10.3390/ijms221910257] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/03/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
Apoptosis, or programmed cell death, is a form of cell suicide that is extremely important for ridding the body of cells that are no longer required, to protect the body against hazardous cells, such as cancerous ones, and to promote tissue morphogenesis during animal development. Upon reception of a death stimulus, the doomed cell activates biochemical pathways that eventually converge on the activation of dedicated enzymes, caspases. Numerous pieces of information on the biochemical control of the process have been gathered, from the successive events of caspase activation to the identification of their targets, such as lamins, which constitute the nuclear skeleton. Yet, evidence from multiple systems now shows that apoptosis is also a mechanical process, which may even ultimately impinge on the morphogenesis of the surrounding tissues. This mechanical role relies on dramatic actomyosin cytoskeleton remodelling, and on its coupling with the nucleus before nucleus fragmentation. Here, we provide an overview of apoptosis before describing how apoptotic forces could combine with selective caspase-dependent proteolysis to orchestrate nucleus destruction.
Collapse
|
24
|
Abramyan J, Geetha-Loganathan P, Šulcová M, Buchtová M. Role of Cell Death in Cellular Processes During Odontogenesis. Front Cell Dev Biol 2021; 9:671475. [PMID: 34222243 PMCID: PMC8250436 DOI: 10.3389/fcell.2021.671475] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/24/2021] [Indexed: 01/20/2023] Open
Abstract
The development of a tooth germ in a precise size, shape, and position in the jaw, involves meticulous regulation of cell proliferation and cell death. Apoptosis, as the most common type of programmed cell death during embryonic development, plays a number of key roles during odontogenesis, ranging from the budding of the oral epithelium during tooth initiation, to later tooth germ morphogenesis and removal of enamel knot signaling center. Here, we summarize recent knowledge about the distribution and function of apoptotic cells during odontogenesis in several vertebrate lineages, with a special focus on amniotes (mammals and reptiles). We discuss the regulatory roles that apoptosis plays on various cellular processes during odontogenesis. We also review apoptosis-associated molecular signaling during tooth development, including its relationship with the autophagic pathway. Lastly, we cover apoptotic pathway disruption, and alterations in apoptotic cell distribution in transgenic mouse models. These studies foster a deeper understanding how apoptotic cells affect cellular processes during normal odontogenesis, and how they contribute to dental disorders, which could lead to new avenues of treatment in the future.
Collapse
Affiliation(s)
- John Abramyan
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, MI, United States
| | | | - Marie Šulcová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
| | - Marcela Buchtová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
| |
Collapse
|
25
|
DNase II mediates a parthanatos-like developmental cell death pathway in Drosophila primordial germ cells. Nat Commun 2021; 12:2285. [PMID: 33863891 PMCID: PMC8052343 DOI: 10.1038/s41467-021-22622-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 03/23/2021] [Indexed: 02/02/2023] Open
Abstract
During Drosophila embryonic development, cell death eliminates 30% of the primordial germ cells (PGCs). Inhibiting apoptosis does not prevent PGC death, suggesting a divergence from the conventional apoptotic program. Here, we demonstrate that PGCs normally activate an intrinsic alternative cell death (ACD) pathway mediated by DNase II release from lysosomes, leading to nuclear translocation and subsequent DNA double-strand breaks (DSBs). DSBs activate the DNA damage-sensing enzyme, Poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) and the ATR/Chk1 branch of the DNA damage response. PARP-1 and DNase II engage in a positive feedback amplification loop mediated by the release of PAR polymers from the nucleus and the nuclear accumulation of DNase II in an AIF- and CypA-dependent manner, ultimately resulting in PGC death. Given the anatomical and molecular similarities with an ACD pathway called parthanatos, these findings reveal a parthanatos-like cell death pathway active during Drosophila development.
Collapse
|